Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Neurol ; 31(1): e16079, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37789648

RESUMEN

BACKGROUND AND PURPOSE: It is not known whether the route of administration affects the mechanisms of action of therapeutic immunoglobulin in chronic inflammatory demyelinating polyneuropathy (CIDP). The aim of this study, therefore, was to compare the immunomodulatory effects of intravenous (IVIg) and subcutaneous immunoglobulin (SCIg) in patients with CIDP and in IVIg-treated common variable immunodeficiency (CVID) patients. METHODS: Serum and peripheral blood mononuclear cell samples were obtained from 30 CIDP patients receiving IVIg, 10 CIDP patients receiving SCIg, and 15 patients with CVID receiving IVIg. Samples and clinical data were obtained prior to IVIg/SCIg and at 3 days, 7 days, and, in CIDP patients receiving IVIg, 21 days post-administration. Serum cytokines were assessed by Luminex-based multiplex assay and enzyme-linked immunosorbent assay. Immune cells were characterized by flow cytometry. RESULTS: Immune cell profiles of CIDP and CVID patients differed in frequencies of myeloid dendritic cells and cytotoxic natural killer cells. During treatment with IVIg or SCIg in CIDP patients, cellular immunomarkers were largely similar. CIDP patients receiving IVIg had higher macrophage inflammatory protein (MIP)-1α (p = 0.01), interleukin (IL)-4 (p = 0.04), and IL-33 (p = 0.04) levels than SCIg recipients. IVIg treatment more broadly modulated cytokines in CIDP than SCIg treatment. CONCLUSIONS: Our study demonstrates that the modulation of cellular immunomarkers in CIDP is independent of the application route of therapeutic immunoglobulin. Minor differences were observed between CIDP and CVID patients. In contrast, cytokines were differentially modulated by IVIg and SCIg in CIDP.


Asunto(s)
Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante , Humanos , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/tratamiento farmacológico , Inmunoglobulinas Intravenosas/uso terapéutico , Leucocitos Mononucleares , Administración Intravenosa , Citocinas
2.
Eur J Neurol ; 29(11): 3380-3388, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35842740

RESUMEN

BACKGROUND AND PURPOSE: This study assessed the prevalence of anti-SARS-CoV-2 antibodies in therapeutic immunoglobulin and their impact on serological response to COVID-19 mRNA vaccine in patients with intravenous immunoglobulin (IVIg)-treated chronic immune neuropathies. METHODS: Forty-six samples of different brands or lots of IVIg or subcutaneous IgG were analyzed for anti-SARS-CoV-2 IgG using enzyme-linked immunosorbent assay and chemiluminescent microparticle immunoassay. Blood sera from 16 patients with immune neuropathies were prospectively analyzed for anti-SARS-CoV-2 IgA, IgG, and IgM before and 1 week after IVIg infusion subsequent to consecutive COVID-19 mRNA vaccine doses and after 12 weeks. These were compared to 42 healthy subjects. RESULTS: Twenty-four (52%) therapeutic immunoglobulin samples contained anti-SARS-CoV-2 IgG. All patients with immune neuropathies (mean age = 65 ± 16 years, 25% female) were positive for anti-SARS-CoV-2 IgG after COVID-19 vaccination. Anti-SARS-CoV-2 IgA titers significantly decreased 12-14 weeks after vaccination (p = 0.02), whereas IgG titers remained stable (p = 0.2). IVIg did not significantly reduce intraindividual anti-SARS-CoV-2 IgA/IgG serum titers in immune neuropathies (p = 0.69). IVIg-derived anti-SARS-CoV-2 IgG did not alter serum anti-SARS-CoV-2 IgG decrease after IVIg administration (p = 0.67). CONCLUSIONS: Our study indicates that IVIg does not impair the antibody response to COVID-19 mRNA vaccine in a short-term observation, when administered a minimum of 2 weeks after each vaccine dose. The infusion of current IVIg preparations that contain anti-SARS-CoV-2 IgG does not significantly alter serum anti-SARS-CoV-2 IgG titers.


Asunto(s)
COVID-19 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales , Formación de Anticuerpos , Vacunas contra la COVID-19 , Femenino , Humanos , Inmunoglobulina A , Inmunoglobulina G , Inmunoglobulina M , Inmunoglobulinas Intravenosas/uso terapéutico , Masculino , Persona de Mediana Edad , SARS-CoV-2 , Vacunación , Vacunas Sintéticas , Vacunas de ARNm
3.
J Neuroinflammation ; 17(1): 33, 2020 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-31980036

RESUMEN

BACKGROUND: In cerebral ischemia, microglia have a dichotomous role in keeping the balance between pro- and anti-inflammatory mediators to avoid deleterious chronic inflammation and to leverage repair processes. METHODS: We examined functional and inflammatory markers in primary rat microglia in vitro after oxygen-glucose deprivation (OGD) or glucose deprivation (aglycemia). We then investigated the preconditioning effect of OGD or aglycemia upon a subsequent strong inflammatory stimulus, here lipopolysaccharides (LPS). Moreover, an "in vitro brain model" of neurons and glia, differentiated from primary rat neural stem cells, was exposed to OGD or aglycemia. Conditioned medium (CM) of this neuronal/glial co-culture was then used to condition microglia, followed by LPS as a "second hit." RESULTS: OGD or aglycemia at sublethal doses did not significantly affect microglia function, including the expression of inflammatory markers. However, preconditioning with either OGD or aglycemia led to a decreased pro-inflammatory response to a subsequent stimulus with LPS. Interestingly, the anti-inflammatory markers IGF-1 and IL-10 were additionally reduced after such preconditioning, while expression of CD206 remained unaffected. Treatment with CM from the neuronal/glial co-culture alone did not affect the expression of inflammatory markers in microglia. In contrast, treatment with CM increased the expression of both pro- and anti-inflammatory markers in microglia upon a second hit with LPS. Interestingly, this effect could be attenuated in microglia treated with CM from neuronal/glia co-cultures preconditioned with OGD or aglycemia. CONCLUSIONS: Data suggest specific and distinct microglia signatures in response to metabolic stress. While metabolic stress directly and indirectly applied to microglia did not mitigate their subsequent response to inflammation, preconditioning with metabolic stress factors such as OGD and aglycemia elicited a decreased inflammatory response to a subsequent inflammation stimulus.


Asunto(s)
Inflamación/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Receptor Cross-Talk/fisiología , Estrés Fisiológico/fisiología , Animales , Isquemia Encefálica/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Glucosa/deficiencia , Inflamación/inducido químicamente , Lipopolisacáridos/farmacología , Ratas
4.
Mediators Inflamm ; 2017: 7189421, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29104378

RESUMEN

BACKGROUND: Focal cerebral ischemia induces distinct neuroinflammatory processes. We recently reported the extracellular phosphor-glyco-protein osteopontin (OPN) to directly affect primary microglia in vitro, promoting survival while shifting their inflammatory profile towards a more neutral phenotype. We here assessed the effects of OPN on microglia after stroke in vivo, with focus on infarct demarcation. METHODS: Animals underwent focal photothrombotic stroke and were injected intracerebroventricularly with 500 µg OPN or vehicle. Immunohistochemistry assessed neuronal damage and infarct volume, neovascularisation, glial scar formation, microglial activation, and M1 and M2 polarisation. RESULTS: After photothrombotic stroke, areas covered by M1 and M2 microglia substantially overlapped. OPN treatment reduced that overlap, with microglia appearing more spread out and additionally covering the infarct core. OPN additionally modulated the quantity of microglia subpopulations, reducing iNOS+ M1 cells while increasing M2 microglia, shifting the M1/M2 balance towards an M2 phenotype. Moreover, OPN polarized astrocytes towards the infarct. CONCLUSION: Microglial activation and M1 and M2 polarization have distinct but overlapping spatial patterns in permanent focal ischemia. Data suggest that OPN is involved in separating M1 and M2 subpopulations, as well as in shifting microglia polarization towards the M2 phenotype modulating beneficially inflammatory responses after focal infarction.


Asunto(s)
Microglía/efectos de los fármacos , Osteopontina/uso terapéutico , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Inmunohistoquímica , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Microglía/metabolismo , Ratas , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo
5.
Neuroimage ; 128: 54-62, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26747749

RESUMEN

Inflammatory cells such as microglia need energy to exert their functions and to maintain their cellular integrity and membrane potential. Subsequent to cerebral ischemia, inflammatory cells infiltrate tissue with limited blood flow where neurons and astrocytes died due to insufficient supply with oxygen and glucose. Using dual tracer positron emission tomography (PET), we found that concomitant with the presence of inflammatory cells, transport and consumption of glucose increased up to normal levels but returned to pathological levels as soon as inflammatory cells disappeared. Thus, inflammatory cells established sufficient glucose supply to satisfy their energy demands even in regions with insufficient supply for neurons and astrocytes to survive. Our data suggest that neurons and astrocytes died from oxygen deficiency and inflammatory cells metabolized glucose non-oxidatively in regions with residual availability. As a consequence, glucose metabolism of inflammatory cells can mask metabolic deficits in neurodegenerative diseases. We further found that the PET tracer did not bind to inflammatory cells in severely hypoperfused regions and thus only a part of the inflammation was detected. We conclude that glucose consumption of inflammatory cells should be taken into account when analyzing disease-related alterations of local cerebral metabolism.


Asunto(s)
Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Inflamación/metabolismo , Animales , Encéfalo/patología , Procesamiento de Imagen Asistido por Computador , Inflamación/patología , Imagen por Resonancia Magnética , Masculino , Tomografía de Emisión de Positrones , Ratas , Ratas Wistar
6.
Mult Scler Relat Disord ; 87: 105664, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38735204

RESUMEN

BACKGROUND: Natalizumab is an effective treatment for relapsing multiple sclerosis (MS). During therapy, individuals are at increased risk of developing progressive multifocal leukoencephalopathy (PML). So far, the relevant reservoir for PML-type JC polyomavirus (JCV) remains elusive. We here tested if the detection of JCV-DNA in stool of persons with MS treated with natalizumab could be a future tool for PML risk assessment. METHODS: The presence of JCV-DNA in stool, urine, and whole blood of MS patients treated with natalizumab and known serum anti-JCV antibodies index values (IV) was studied. Different DNA extraction methods, real-time (RT) and droplet digital (dd) PCR techniques were compared. JCV isolates were screened for PML-associated variants by sequencing. RESULTS: Thirty MS patients treated with natalizumab were screened. For 21 patients, blood, stool, and urine samples were available. These patients were stratified according to their serum anti-JCV antibody IV (high (>1.5, n = 12); medium (1.5-0.9, n = 2); low (<0.9, n = 1); negative (n = 6)). JCV-DNA could not be detected in the whole blood or stool samples. Four urine samples had measurable JCV-DNA, ranging from 1.71×104-1.07×108 international units (IU)/mL detected by RT-PCR, corresponding to 4.62×104-9.85×106 copies/mL measured by ddPCR. All JCV variants were wild-type and derived from patients with high antibody IV. CONCLUSION: Stool-specific DNA extraction methods provided the highest quality of DNA, while the sensitivity of ddPCR and RT- PCR was comparable. Our findings do not support assessing stool samples for PML risk stratification in persons with MS. Further studies are needed to explore where PML-associated viral variants arise.


Asunto(s)
Anticuerpos Antivirales , ADN Viral , Heces , Factores Inmunológicos , Virus JC , Natalizumab , Humanos , Virus JC/aislamiento & purificación , Virus JC/inmunología , Natalizumab/uso terapéutico , Heces/virología , Adulto , Masculino , Femenino , Anticuerpos Antivirales/sangre , ADN Viral/sangre , ADN Viral/análisis , Persona de Mediana Edad , Leucoencefalopatía Multifocal Progresiva/sangre , Leucoencefalopatía Multifocal Progresiva/virología , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/virología , Esclerosis Múltiple/sangre
7.
Fluids Barriers CNS ; 17(1): 65, 2020 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-33109245

RESUMEN

BACKGROUND: Progressive multifocal leukoencephalopathy (PML) is caused by an opportunistic infection with JC polyoma virus (JCPyV) and mainly affects immunocompromised patients. It leads to pronounced demyelination of the central nervous system (CNS) resulting in severe disability or even death. Detection of JCPyV DNA in the cerebrospinal fluid (CSF) is usually accepted as proof for the diagnosis of PML. Routine CSF parameters, like CSF cell count, protein concentration, Qalbumin, or intrathecal immunoglobulin synthesis are mostly considered normal. However, this has not been investigated systematically. METHODS: We analyzed routine CSF parameters in a cohort of 108 PML patients that were treated at four different neurological centers in Germany. The patients exhibited different underlying conditions with natalizumab-treated multiple sclerosis (n = 54) and human immunodeficiency virus (HIV)-infection (n = 25) being the most frequent. The data were collected at the respective centers in accordance with local requirements and then jointly analyzed. The total PML cohort was compared with a control group of patients with normal pressure hydrocephalus (NPH) and idiopathic intracranial hypertension (IIH). Multiple sclerosis and HIV patients were additionally compared with their own non-PML control groups. RESULTS: The PML group showed an elevated cell count (p < 0.001) compared to the control group, however, this effect was mainly driven by HIV-PML patients. This subgroup also demonstrated a significantly higher proportion of patients with a disturbed blood-CSF-barrier function. CONCLUSIONS: This comprehensive, retrospective study on CSF diagnostic analysis in PML patients provides insight into the CSF of those patients. It demonstrates that CSF composition in PML patients may be specific for the underlying condition that predisposes for the development of PML and thus data have to be interpreted in this context.


Asunto(s)
Líquido Cefalorraquídeo/citología , Líquido Cefalorraquídeo/metabolismo , Infecciones por VIH/líquido cefalorraquídeo , Leucoencefalopatía Multifocal Progresiva/líquido cefalorraquídeo , Esclerosis Múltiple/líquido cefalorraquídeo , Adulto , Femenino , Infecciones por VIH/complicaciones , Humanos , Hidrocéfalo Normotenso/líquido cefalorraquídeo , Leucoencefalopatía Multifocal Progresiva/etiología , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones , Seudotumor Cerebral/líquido cefalorraquídeo , Estudios Retrospectivos
8.
J Neuroimmune Pharmacol ; 14(2): 295-311, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30488353

RESUMEN

Cortical cerebral ischemia elicits neuroinflammation as well as secondary neuronal degeneration in remote areas. Locally distinct and specific secondary neurodegeneration affecting thalamic nuclei connected to cortical areas highlights such processes. Osteopontin (OPN) is a cytokine-like glycoprotein that is excreted in high amounts after cerebral ischemia and exerts various immunomodulatory functions. We here examined putative protective effects of OPN in secondary thalamic degeneration. We subjected male Wistar rats to photothrombosis and subsequently injected OPN or placebo intracerebroventricularly. Immunohistochemical and fluorescence staining was used to detect the extent of neuronal degeneration and microglia activation. Ex vivo autoradiography with radiotracers available for human in vivo PET studies, i.e., CIS-4-[18F]Fluor-D-Proline (D-cis-[18F]FPRO), and [6-3H]thymidine ([3H]thymidine), confirmed degeneration and proliferation, respectively. We found secondary neurodegeneration in the thalamus characterized by microglial activation and neuronal loss. Neuronal loss was restricted to areas of microglial infiltration. Treatment with OPN significantly decreased neurodegeneration, inflammation and microglial proliferation. Microglia displayed morphological signs of activation without expressing markers of M1 or M2 polarization. D-CIS-[18F]FPRO-uptake mirrored attenuated degeneration in OPN-treated animals. Notably, [3H]thymidine and BrdU-staining revealed increased stem cell proliferation after treatment with OPN. The data suggest that OPN is able to ameliorate secondary neurodegeneration in thalamic nuclei. These effects can be visualized by radiotracers D-CIS-[18F]FPRO and [3H]thymidine, opening new vistas for translational studies. Graphical Abstract Intracerebroventricular injection of osteopontin attenuates thalamic degeneration after cortical ischemia (pink area). Disruption of thalamocortical connections (blue) and degeneration of thalamic nuclei (encircled) leads to microglia activation. Osteopontin protects from both neurodegeneration and microglia activation as assessed by histological analysis and autoradiograpic studies.


Asunto(s)
Enfermedades Neurodegenerativas/prevención & control , Osteopontina/farmacología , Accidente Cerebrovascular/patología , Enfermedades Talámicas/prevención & control , Tálamo/patología , Animales , Isquemia Encefálica/patología , Activación de Macrófagos/efectos de los fármacos , Masculino , Microglía/efectos de los fármacos , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Fagocitos/efectos de los fármacos , Fagocitos/patología , Tomografía de Emisión de Positrones , Ratas , Ratas Wistar , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/diagnóstico por imagen , Enfermedades Talámicas/diagnóstico por imagen , Enfermedades Talámicas/patología , Tálamo/diagnóstico por imagen , Trombosis/patología
9.
Front Cell Neurosci ; 13: 461, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708742

RESUMEN

Despite its extensive use in clinical studies, the molecular mechanisms underlying the effects of transcranial direct current stimulation (tDCS) remain to be elucidated. We previously described subacute effects of tDCS on immune- and stem cells in the rat brain. To investigate the more immediate effects of tDCS regulating those cellular responses, we treated rats with a single session of either anodal or cathodal tDCS, and analyzed the gene expression by microarray; sham-stimulated rats served as control. Anodal tDCS increased expression of several genes coding for the major histocompatibility complex I (MHC I), while cathodal tDCS increased the expression of the immunoregulatory protein osteopontin (OPN). We confirmed the effects of gene upregulation by immunohistochemistry at the protein level. Thus, our data show a novel mechanism for the actions of tDCS on immune- and inflammatory processes, providing a target for future therapeutic studies.

10.
J Neuroimmune Pharmacol ; 11(4): 708-720, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27352075

RESUMEN

The neural cell adhesion molecule (NCAM)-derived peptide FG loop (FGL) modulates synaptogenesis, neurogenesis, and stem cell proliferation, enhances cognitive capacities, and conveys neuroprotection after stroke. Here we investigated the effect of subcutaneously injected FGL on cellular compartments affected by degeneration and regeneration after stroke due to middle cerebral artery occlusion (MCAO), namely endogenous neural stem cells (NSC), oligodendrocytes, and microglia. In addition to immunohistochemistry, we used non-invasive positron emission tomography (PET) imaging with the tracer [18F]-fluoro-L-thymidine ([18F]FLT) to visualize endogenous NSC in vivo. FGL significantly increased endogenous NSC mobilization in the neurogenic niches as evidenced by in vivo and ex vivo methods, and it induced remyelination. Moreover, FGL affected neuroinflammation. Extending previous in vitro results, our data show that the NCAM mimetic peptide FGL mobilizes endogenous NSC after focal ischemia and enhances regeneration by amplifying remyelination and modulating neuroinflammation via affecting microglia. Results suggest FGL as a promising candidate to promote recovery after stroke.


Asunto(s)
Movimiento Celular/fisiología , Regeneración Nerviosa/fisiología , Moléculas de Adhesión de Célula Nerviosa/administración & dosificación , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Péptidos/administración & dosificación , Accidente Cerebrovascular/patología , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Inyecciones Subcutáneas , Masculino , Regeneración Nerviosa/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Tomografía de Emisión de Positrones/tendencias , Ratas , Ratas Wistar , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/tratamiento farmacológico
11.
Exp Neurol ; 279: 127-136, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26923911

RESUMEN

BACKGROUND: Clinical data suggest that transcranial direct current stimulation (tDCS) may be used to facilitate rehabilitation after stroke. However, data are inconsistent and the neurobiological mechanisms underlying tDCS remain poorly explored, impeding its implementation into clinical routine. In the healthy rat brain, tDCS affects neural stem cells (NSC) and microglia. We here investigated whether tDCS applied after stroke also beneficially affects these cells, which are known to be involved in regeneration and repair. METHODS: Focal cerebral ischemia was induced in rats by transient occlusion of the middle cerebral artery. Twenty-eight animals with comparable infarcts, as judged by magnetic resonance imaging, were randomized to receive a multi-session paradigm of either cathodal, anodal, or sham tDCS. Behaviorally, recovery of motor function was assessed by Catwalk. Proliferation in the NSC niches was monitored by Positron-Emission-Tomography (PET) employing the radiotracer 3'-deoxy-3'-[(18)F]fluoro-l-thymidine ([(18)F]FLT). Microglia activation was depicted with [(11)C]PK11195-PET. In addition, immunohistochemical analyses were used to quantify neuroblasts, oligodendrocyte precursors, and activation and polarization of microglia. RESULTS: Anodal and cathodal tDCS both accelerated functional recovery, though affecting different aspects of motor function. Likewise, tDCS induced neurogenesis independently of polarity, while only cathodal tDCS recruited oligodendrocyte precursors towards the lesion. Moreover, cathodal stimulation preferably supported M1-polarization of microglia. CONCLUSIONS: TDCS acts through multifaceted mechanisms that far exceed its primary neurophysiological effects, encompassing proliferation and migration of stem cells, their neuronal differentiation, and modulation of microglia responses.


Asunto(s)
Células-Madre Neurales , Neurogénesis , Oligodendroglía , Accidente Cerebrovascular/terapia , Estimulación Transcraneal de Corriente Directa/métodos , Animales , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Electrodos , Infarto de la Arteria Cerebral Media/patología , Activación de Macrófagos , Masculino , Microglía , Regeneración Nerviosa , Tomografía de Emisión de Positrones , Desempeño Psicomotor , Ratas , Ratas Wistar , Recuperación de la Función , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/patología
12.
Stem Cell Res Ther ; 6: 99, 2015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-25998490

RESUMEN

INTRODUCTION: Osteopontin (OPN) is a phosphoglycoprotein with important roles in tissue homeostasis, wound healing, immune regulation, and stress responses. It is expressed constitutively in the brain and upregulated during neuroinflammatory responses; for example, after focal cerebral ischemia. To date, its effects on neural stem cells (NSC) remain to be elucidated and are, accordingly, the subject of this study. METHOD: Primary fetal rat NSC were cultured as homogenous monolayers and treated with different concentrations of OPN. Fundamental properties of NSC were assessed following OPN exposure, including proliferative activity, survival under oxidative stress, migration, and differentiation potential. To elucidate a putative action of OPN via the CXC chemokine receptor type 4 (CXCR4), the latter was blocked with AMD3100. To investigate effects of OPN on endogenous NSC in vivo, recombinant OPN was injected into the brain of healthy adult rats as well as rats subjected to focal cerebral ischemia. Effects of OPN on NSC proliferation and neurogenesis in the subventricular zone were studied immunohistochemically. RESULTS: OPN dose-dependently increased the number of NSC in vitro. As hypothesized, this effect was mediated through CXCR4. The increase in NSC number was due to both enhanced cell proliferation and increased survival, and was confirmed in vivo. Additionally, OPN dose-dependently stimulated the migration of NSC via CXCR4. Moreover, in the presence of OPN, differentiation of NSC led to a significant increase in neurogenesis both in vitro as well as in vivo after cerebral ischemia. CONCLUSION: Data show positive effects of OPN on survival, proliferation, migration, and neuronal differentiation of NSC. At least in part these effects were mediated via CXCR4. Results suggest that OPN is a promising substance for the targeted activation of NSC in future experimental therapies for neurological disorders such as stroke.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células-Madre Neurales/metabolismo , Osteopontina/farmacología , Receptores CXCR4/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Feto/citología , Inmunohistoquímica , Células-Madre Neurales/citología , Neurogénesis/efectos de los fármacos , Ratas , Transducción de Señal/efectos de los fármacos
13.
Neuron ; 84(6): 1183-90, 2014 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-25433639

RESUMEN

Impaired axonal transport can contribute to axon degeneration and has been described in many neurodegenerative diseases. Multiple sclerosis (MS) is a common neuroinflammatory disease, which is characterized by progressive axon degeneration-whether, when, and how axonal transport is affected in this condition is unknown. Here we used in vivo two-photon imaging to directly assay transport of organelles and the stability of microtubule tracks in individual spinal axons in mouse models of MS. We found widespread transport deficits, which preceded structural alterations of axons, cargos, or microtubules and could be reversed by acute anti-inflammatory interventions or redox scavenging. Our study shows that acute neuroinflammation induces a pervasive state of reversible axonal dysfunction, which coincides with acute disease symptoms. Moreover, perpetuated transport dysfunction, as we found in a model of progressive MS, led to reduced distal organelle supply and could thus contribute to axonal dystrophy in advanced stages of the disease.


Asunto(s)
Transporte Axonal/fisiología , Axones/fisiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Animales , Transporte Axonal/efectos de los fármacos , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/farmacología , Neuroimagen Funcional , Ratones , Microtúbulos/fisiología , Degeneración Nerviosa/fisiopatología , Donantes de Óxido Nítrico/farmacología , Orgánulos/fisiología , Espermina/análogos & derivados , Espermina/farmacología , Médula Espinal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA